EP3034625A1 - Procédé ultrasensible de détection in situ d'acides nucléiques - Google Patents

Procédé ultrasensible de détection in situ d'acides nucléiques Download PDF

Info

Publication number
EP3034625A1
EP3034625A1 EP15194058.2A EP15194058A EP3034625A1 EP 3034625 A1 EP3034625 A1 EP 3034625A1 EP 15194058 A EP15194058 A EP 15194058A EP 3034625 A1 EP3034625 A1 EP 3034625A1
Authority
EP
European Patent Office
Prior art keywords
probe
label
nucleic acid
cell
signal amplification
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP15194058.2A
Other languages
German (de)
English (en)
Other versions
EP3034625B1 (fr
Inventor
Xingyong Wu
Huei-Yu Fay Wang
Nan Su
Li-Chong Wang
Yuling Luo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Advanced Cell Diagnostics Inc
Original Assignee
Advanced Cell Diagnostics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Advanced Cell Diagnostics Inc filed Critical Advanced Cell Diagnostics Inc
Publication of EP3034625A1 publication Critical patent/EP3034625A1/fr
Application granted granted Critical
Publication of EP3034625B1 publication Critical patent/EP3034625B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • C12Q1/682Signal amplification
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • C12Q1/6818Hybridisation assays characterised by the detection means involving interaction of two or more labels, e.g. resonant energy transfer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6841In situ hybridisation

Definitions

  • the invention relates generally to nucleic acid chemistry and biochemical assays. More particularly, the invention relates to methods for in situ detection of nucleic acid analytes in a sample.
  • ISH In situ hybridization
  • ISH tissue-specific and cell-type specific express patterns together with expression level of the gene can be obtained with ISH, which will provide valuable information for analyzing the function of the gene.
  • RNAscope® a new ISH signal amplification method called Advanced Cell Diagnostics, Inc. ( US Patent No. 7,709,198 ). This assay includes uniquely designed oligo capture probes and a signal amplification system composed of preamplifiers, amplifiers, and label probes, allowing substantial signal amplification without amplifying background signal, enabling single RNA molecule detection for virtually any genes.
  • An exemplary embodiment of the RNAscope® technology is schematically illustrated in Fig.1 and will be described in detail in Section 2 of this application.
  • RNAscope® In a typical RNAscope® assay for detecting target nucleic acid, a target mRNA whose expression is to be detected is released from cells and captured on a solid surface (e.g. , a well of a microtiter plate). A set of two or more capture probes and a signal generating multimer are also provided. The capture probes hybridize to both target nucleic acid and signal generating multimer and thus capture signal generating multimer to target nucleic acid.
  • the signal generating multimer comprises label probes (LPs). But more typically, the signal generating multimer comprises preamplifiers and/or amplifiers in addition to label probes.
  • the label probe is capable of binding to a label particle or molecule that provide detectable signal.
  • the label probe has a larger molecular structure enabling that attachment of a plurality of label particle or molecules that provide stronger signal than a single label particle or molecule. Thus, RNAscope® improves the sensitivity and specific of nucleic acid detection.
  • RNAscope® alone still cannot reliably detect some low copy genes in retrospective Formalin-Fixed, Paraffin-Embedded (FFPE) tissue sections where RNA is significantly degraded. Also, single RNA molecule cannot to be visualized at 40X magnification using current RNAscope® technology. It is desirable to further enhance the detection signal to enable more robust detection of any RNA molecules, including the significantly degraded RNA molecules, and to allow easy visualization of detected RNA signal at 10X magnification.
  • FFPE Paraffin-Embedded
  • RNAscope® in combination with biotin-(strept)avidin
  • RNAscope® in combination with an antibody
  • TSA Tyramide Signal Amplification
  • Biotin-avidin (or biotin-streptavidin) is a well known signal amplification system base on the facts that the two molecules have extraordinarily high affinity to each other and that one avidin/streptavidin molecule can bind four biotin molecules.
  • Antibodies are widely used for signal amplification in immunohistochemistry and ISH.
  • TSA is based on the deposition of a large number of haptenized tyramide molecules by peroxidase activity. Tyramine is a phenolic compound.
  • immobilized Horse Radish Peroxidase (HRP) converts the labeled substrate into a short-lived, extremely reactive intermediate.
  • the activated substrate molecules then very rapidly react with and covalently bind to electron-rich moieties of proteins, such as tyrosine, at or near the site of the peroxidase binding site. In this way, a lot of extra hapten molecules conjugated to tyramide can be introduced at the hybridization site in situ. Subsequently, the deposited tyramide-hapten molecules can be visualized directly or indirectly.
  • proteins such as tyrosine
  • the present invention combines the signal amplification method of RNAscope® with the general ISH signal amplification methods into a ultra sensitive method to detect, quantify and identify one or more target nucleic acid in a sample.
  • the RNAscope® assay offers exceptional specificity owning to its unique paired capture probe design.
  • general ISH signal amplification methods such as the TSA-based signal amplification
  • the combined method achieves great signal intensity and low background.
  • the signal amplification methods and system disclosed in this invention are relatively simple to use and produce consistent result. The claimed method can be easily adapted for use in routine clinic diagnostic procedures.
  • the specificity and sensitivity of the disclosed signal amplification method are balanced. The balance is achieved by a moderate reduction of the fold of signal amplification in RNAscope®.
  • one preamplifer is designed to bind between 1 to 16 amplifiers. In a preferred embodiment, one preamplifer is designed to bind between 2 to 10 amplifiers. In another more preferred embodiment, one preamplifer is designed to bind between 2 to 5 amplifiers.
  • the sample comprising or suspected of comprising the target nucleic acid is first provided, together with at least one set of two or more capture probes capable of hybridizing to the target nucleic acid, a signal generating multimer capable of hybridizing to the set of two or more capture probes, wherein said signal generating multimer comprises a label probe, and a signal amplification probe capable of binding to the label probe wherein the signal amplification probe comprises a label.
  • the target nucleic acid is first hybridized to the set of two or more capture probes, then the signal generating multimer is captured to the set of two or more capture probes and thereby being captured the signal generating multimer to the target nucleic acid.
  • the signal amplification probe is then captured to the label probe and thereby being captured to the signal generating multimer.
  • the presence, absence, or amount of the label in the signal amplification probe are detected.
  • the signal amplification probe comprises: a biotin molecule which is capable of conjugating to the label probe, an avidin/streptavidin molecule which is capable of binding to the biotin molecule, and additional biotin molecules being conjugated to Horse Radish Peroxidase (HRP), Alkaline Phosphatase (AP), or fluorephore and are capable of binding to the avidin/streptavidin molecule.
  • HRP Horse Radish Peroxidase
  • AP Alkaline Phosphatase
  • fluorephore fluorephore
  • the signal amplification probe comprises: a HRP, AP, Dinitrophenyl (DNP), or fluorophore molecule which is capable of conjugating to the label probe, one or more first antibody which is capable of binding to said HRP, AP, DNP, or fluorophore molecule, and one or more second antibody being conjugated to HRP, Polymer-HRP, AP, Polymer-AP or fluorophore and is capable of binding to said one or more first antibody.
  • a HRP, AP, Dinitrophenyl (DNP), or fluorophore molecule which is capable of conjugating to the label probe
  • one or more first antibody which is capable of binding to said HRP, AP, DNP, or fluorophore molecule
  • second antibody being conjugated to HRP, Polymer-HRP, AP, Polymer-AP or fluorophore and is capable of binding to said one or more first antibody.
  • the signal amplification probe comprises: a HRP molecule which is capable of conjugating to said label probe, a plurality of tyramide-biotin or tyramide-fluorophore molecules which are capable of reacting with said HRP molecule, and detection labels which are capable detecting said tyramide-biotin or tyramide-fluorophore molecules visually.
  • the detection labels are a combination of avidin/streptavidin-HRP and a chromogenic substrate or a combination of avidin/streptavidin-AP and a chromogenic substrate.
  • the chromogenic substrate is selected from the group consisting of: diaminobenzine(DAB) and Fast Red.
  • the signal generating multimer comprises a label probe capable of hybridizing to the set of two or more capture probes. In another embodiment, the signal generating multimer comprises a label probe and an amplifier hybridized to the label probe. The amplifier is capable of hybridizing to said set of two or more capture probes. In a preferred embodiment, the signal generating multimer comprises a label probe, an amplifier hybridized to the label, and a preamplifier hybridized to one or more of the amplifier. The preamplifier is capable of hybridizing to said set of two or more capture probes.
  • the target nucleic acid can be any type, such as, e..g, DNA, cDNA, RNA, mRNA, rRNA, miRNA, siRNA and/or the like.
  • the amplification can take place on a solid support before hybridization is performed.
  • Methods of the invention further provide a variety of ways to multiplex two or more target nucleic acids.
  • the sample may comprise a cell comprising or suspected of comprising two or more different target nucleic acids.
  • the sample may also comprise two or more different cells, each comprising or suspected of comprising a different target nucleic acid.
  • the two different target nucleic acids can be detected using a dual-color chromogenic in situ hybridization (CISH) or a dual-color fluorescent in situ hybridization (FISH).
  • CISH dual-color chromogenic in situ hybridization
  • FISH dual-color fluorescent in situ hybridization
  • the dual-color CISH is performed using two different signal amplification probes, wherein the first signal amplification probe comprises: tyramide-biotin, streptavidin-HRP and DAB, and the second signal amplification probe comprises: anti-DNP-AP and Fast Red.
  • kits to practice the methods of the invention can include reagents to carry out the nucleic acid detection and provide conditions necessary to practice the methods of the invention.
  • the kits include, e.g. , A kit comprising: a target nucleic acid, one set of two or more capture probes capable of hybridizing to said target nucleic acid, a signal generating multimer capable of hybridizing to the set of two or more capture probes wherein the signal generating multimer comprises a label probe, and a signal amplification probe capable of binding to the label probe, wherein the signal amplification probe comprises a label.
  • the signal amplification probe comprises: a biotin molecule which is capable of conjugating to said label probe, an avidin/streptavidin molecule which is capable of binding to said biotin molecule, and additional biotin molecules being conjugated to HRP, AP or fluorophores and are capable of binding to the avidin/streptavidin molecule.
  • the signal amplification probe comprises: a HRP, AP, DNP, or fluorophore molecule which is capable of conjugating to the label probe, one or more first antibody which is capable of binding to the HRP, AP, DNP, or fluorophore molecule, and one or more second antibody being conjugated to HRP, Polymer-HRP, AP, Polymer-AP or fluorophore and is capable of binding to the one or more first antibody.
  • the signal amplification probe comprises: a HRP molecule which is capable of conjugating to the label probe, a plurality of tyramide-biotin or tyramide-fluorophoremolecules which are capable of reacting with said HRP molecule, and detection labels which are capable detecting the tyramide-biotin molecules visually.
  • the detection labels may be a combination of avidin/streptavidin-HRP and a chromogenic substrate or a combination of avidin/streptavidin-AP and a chromogenic substrate.
  • the chromogenic substrate may be selected from the group consisting of: DAB and Fast Red.
  • the signal generating multimer comprises a label probe capable of hybridizing to the set of two or more capture probes.
  • the signal generating multimer comprises a label probe and an amplifier hybridized to the label probe.
  • the amplifier is capable of hybridizing to said set of two or more capture probes.
  • the signal generating multimer comprises a label probe, an amplifier hybridized to the label, and a preamplifier hybridized to one or more of the amplifier.
  • the preamplifier:amplifier ratio in the kit may be between 1-16, or preferably between 2-10 or between 2-5.
  • the kit of the invention can function in the multiplexing of multiple nucleic acid amplifications.
  • the kit may be used for detecting nucleic acid targets in two or more different cells, each comprising or suspected of comprising a different target nucleic acid, or to detect two or more nucleic acid targets in one cell.
  • the signal amplification probe in the kit may be either a dual-color chromogenic in situ hybridization (CISH) probe or a dual-color fluorescent in situ hybridization (FISH) probe.
  • CISH dual-color chromogenic in situ hybridization
  • FISH fluorescent in situ hybridization
  • the first signal amplification probe comprises: tyramide-biotin, streptavidin-HRP and DAB
  • the second signal amplification probe comprises: anti-DNP-AP and Fast Red.
  • nucleic acid encompasses any physical string of monomer units that can be corresponded to a string of nucleotides, including a polymer of nucleotides (e.g. , a typical DNA or RNA polymer), peptide nucleic acids (PNAs), modified oligonucleotides (e.g. , oligonucleotides comprising nucleotides that are not typical to biological RNA or DNA, such as 2'-O-methylated oligonucleotides), and the like.
  • PNAs peptide nucleic acids
  • modified oligonucleotides e.g. , oligonucleotides comprising nucleotides that are not typical to biological RNA or DNA, such as 2'-O-methylated oligonucleotides
  • the nucleotides of the polynucleotide can be deoxyribonucleotides, ribonucleotides or nucleotide analogs, can be natural or non-natural, and can be unsubstituted, unmodified, substituted or modified.
  • the nucleotides can be linked by phosphodiester bonds, or by phosphorothioate linkages, methylphosphonate linkages, boranophosphate linkages, or the like.
  • the polynucleotide can additionally comprise non-nucleotide elements such as labels, quenchers, blocking groups, or the like.
  • the polynucleotide can be, e.g ., single-stranded or double-stranded.
  • nucleic acid target or “target nucleic acid” refers to a nucleic acid, or optionally a region thereof, that is to be detected.
  • a "polynucleotide sequence” or “nucleotide sequence” is a polymer of nucleotides (an oligonucleotide, a DNA, a nucleic acid, etc.) or a character string representing a nucleotide polymer, depending on context. From any specified polynucleotide sequence, either the given nucleic acid or the complementary polynucleotide sequence (e.g. , the complementary nucleic acid) can be determined.
  • gene is used broadly to refer to any nucleic acid associated with a biological function. Genes typically include coding sequences and/or the regulatory sequences required for expression of such coding sequences. The term gene can apply to a specific genomic sequence, as well as to a cDNA or an mRNA encoded by that genomic sequence.
  • antibody is used herein in the broadest sense and covers fully assembled antibodies, antibody fragments which retain the ability to specifically bind to the antigen (e.g. , Fab, F(ab')2, Fv, and other fragments), single chain antibodies, diabodies, antibody chimeras, hybrid antibodies, bispecific antibodies, humanized antibodies, and the like.
  • antibody covers both polyclonal and monoclonal antibodies.
  • biological sample refers to a sample obtained from a biological subject, including sample of biological tissue or fluid origin, obtained, reached, or collected in vivo or in situ .
  • a biological sample also includes samples from a region of a biological subject containing precancerous or cancer cells or tissues. Such samples can be, but are not limited to, organs, tissues, fractions and cells isolated from a mammal. Exemplary biological samples include but are not limited to cell lysate, a cell culture, a cell line, a tissue, an organ, an organelle, a biological fluid, and the like. Preferred biological samples include but are not limited to a skin sample, tissue biopsies, and the like.
  • label probe refers to an entity that binds to a target molecule, directly or indirectly, and enables the target to be detected, e.g ., by a readout instrument.
  • a label probe (or "LP") is typically a single-stranded polynucleotide that comprises one or more label which directly or indirectly provides a detectable signal.
  • the label can be covalently attached to the polynucleotide, or the polynucleotide can be configured to bind to the label (e.g. , a biotinylated polynucleotide can bind a streptavidin-associated label).
  • the label probe can, for example, hybridize directly to a target nucleic acid, or it can hybridize to a nucleic acid that is in turn hybridized to the target nucleic acid or to one or more other nucleic acids that are hybridized to the nucleic acid.
  • the label probe can comprise a polynucleotide sequence that is complementary to a polynucleotide sequence of the target nucleic acid, or it can comprise at least one polynucleotide sequence that is complementary to a polynucleotide sequence in a capture probe, amplifier, or the like.
  • label is a moiety that facilitates detection of a molecule.
  • Common labels in the context of the present invention include fluorescent, luminescent, light-scattering, and/or colorimetric labels.
  • Suitable labels include enzymes and fluorescent moieties, as well as radionuclides, substrates, cofactors, inhibitors, chemiluminescent moieties, magnetic particles, and the like.
  • Exemplary labels include: Horse Radish Peroxidase (HRP), Alkaline Phosphatase (AP), fluorophore, Dinitrophenyl (DNP), etc. Patents teaching the use of such labels include U.S. Pat. Nos.
  • a “capture probe” is a polynucleotide that is capable of hybridizing to a target nucleic acid and capturing a label probe to that target nucleic acid.
  • the target probe can hybridize directly to the label probe, or it can hybridize to one or more nucleic acids that in turn hybridize to the label probe; for example, the target probe can hybridize to an amplifier or a preamplifier.
  • the target probe thus includes a first polynucleotide sequence that is complementary to a polynucleotide sequence of the target nucleic acid and a second polynucleotide sequence that is complementary to a polynucleotide sequence of the label probe, amplifier, preamplifier, or the like.
  • the target probe is preferably single-stranded.
  • An "amplifier” is a molecule, typically a polynucleotide, that is capable of hybridizing to multiple label probes. Typically, the amplifier hybridizes to multiple identical label probes. The amplifier also hybridizes to at least one target probe or nucleic acid bound to a target probe. For example, the amplifier can hybridize to at least one target probe and to a plurality of label probes, or to a preamplifier and a plurality of label probes. The amplifier can be, e.g ., a linear, forked, comb-like, or branched nucleic acid.
  • the amplifier can include modified nucleotides and/or nonstandard internucleotide linkages as well as standard deoxyribonucleotides, ribonucleotides, and/or phosphodiester bonds. Suitable amplifiers are described, for example, in USPN 5,635,352, USPN 5,124,246, USPN 5,710,264, and USPN 5,849,481.
  • a "preamplifier” is a molecule, typically a polynucleotide, that serves as an intermediate between one or more target probes and amplifiers. Typically, the preamplifier hybridizes simultaneously to one or more target probes and to a plurality of amplifiers. Exemplary preamplifiers are described, for example, in USPN 5,635,352 and USPN 5,681,697.
  • ISH in situ hybridization
  • probe a labeled complementary DNA or RNA strand
  • the probe types are double stranded DNA (dsDNA), single stranded DNA (ssDNA), single stranded complimentary RNA (sscRNA), messenger RNA (mRNA), micro RNA (miRNA), and synthetic oligonucleotides.
  • FISH fluorescent in situ hybridization
  • CISH or "chromogenic in situ hybridization” refers to a type of ISH with a chromogenic label.
  • a "general ISH signal amplification" assay or system refers to any ISH based in situ hybridization assay or system that can amplify the specific DNA or RNA sequence targeted by the method. It includes, but not limited to the three exemplary general ISH signal amplification systems disclosed in the present invention (i.e., biotin-based ISH, antibody-based ISH, and TSA-based ISH).
  • the present invention provides methods and systems for detecting the presence of a target nucleic acid with improved sensitivity and specificity in a sample.
  • the invention combines RNAscope® method with general ISH signal amplification methods such as the TSA-based signal amplification method, the antibody-based signal amplification method, or the Biotin-Avidin-based signal amplification method.
  • the RNAscope® method is known for its high specificity in detecting a target nucleic acid. In other words, a target nucleic acid can be detected with high signal-to-noise ratio in an RNAscope® assay.
  • Such high specificity as will be explained in Section 2 below, are derived from the "double Z" probe design and the preamplifier-amplifier-label probe signal cascade design.
  • General ISH signal amplification methods are known for their ability to boosting signal amplification of nucleic acid molecules. However, ISH signal amplification methods are also known for their high signal to noise ratio and their lack of consistency in the amplification
  • the present invention combines the RNAscope® method with general ISH signal amplification methods and further adjust the amplification ratio between the amplifier and preamplifier in RNAscope®.
  • methods and systems of the present invention overcome the above noted disadvantages of the general ISH signal amplification methods while retaining the unique mechanisms for background noise reduction in RNAscope®.
  • the present invention is capable of reliably detecting nucleic acid targets with great sensitivity and specificity. Such enhancement increases the consistency among the results of nucleic acid detection to the level that can be implemented in diagnostic assays.
  • This invention describes a novel, simple and ultra sensitive in suit hybridization method for detecting target nucleic acid in a sample.
  • This method can be used in both fluorescent ISH (FISH) and chromogenic ISH (CISH) assays.
  • FISH fluorescent ISH
  • CISH chromogenic ISH
  • the method is capable of detecting nuclear acids (DNA and RNA) immobilized in intact cells, tissue sections, tissue microarrays and cDNA microarrays.
  • ISH signal amplification methods There are many ISH signal amplification methods which have been developed to detect DNA or RNA target. Three exemplary ISH signal amplification methods will be described below: (1) Biotin-(Strept)avidin-based ISH signal amplification; (2) Antibody-based ISH signal amplification, and (3) Tyramide Signal Amplification (TSA)-based ISH signal amplification. It is clear to a skilled artisan that any ISH signal amplification method can be used in the present invention in combination with the RNAscope®. Thus, these examples should be not considered as a limitation to the present invention.
  • the biotin-(strept)avidin-based ISH signal amplification method was used in combination with RNAscope®.
  • the RNAscope comprises a set of two or more capture probes, a plurality of label probes, a plurality of amplifiers, and a plurality of preamplifiers.
  • the target nucleic acid is hybridized to the set of capture probes.
  • the set of capture probes is then hybridized to a preamplifier.
  • the preamplifier is further hybridized to a plurality of amplifiers. Each amplifier is then hybridized to a plurality of label probes.
  • the label probe (LP) of the RNAscope complex structure is then conjugated to a biotin molecule of the ISH signal amplification structure and forms a LP-biotin complex.
  • the LP-biotin complex is further bound by a avidin/streptavidin molecule.
  • the avidin/streptavidin molecule has the capacity to bind a maximum of 4 biotin molecules where each biotin molecule is further conjugated to HRP or AP.
  • HRP or AP can be detected visually by chromogenic dyes such as diaminobenzidine, DAB or Fast Red.
  • the antibody-based ISH signal amplification method was used in combination with RNAscope®.
  • the RNAscope comprises a set of two or more capture probes, a plurality of label probes, a plurality of amplifiers, and a plurality of preamplifiers.
  • the target nucleic acid is hybridized to the set of capture probes.
  • the two or more capture probes are hybridized to the nucleic acid target. Every preamplifier are hybridized to each of the two or more capture probes, a plurality of amplifiers is hybridized to each of the preamplifiers, and the plurality of label probes is hybridized to the amplifiers.
  • Each label probe of the RNAscope complex structure is conjugated to an HRP molecule, an AP molecule, a DNP molecule, or a fluorophore molecule and thus forming an HRP-LP complex, an AP-LP complex, a DNP-LP complex, or a fluorophore-LP complex, respectively.
  • the HRP-LP, AP-LP, DNP-LP, or fluorophore-LP complex is recognized by an anti-HRP antibody, anti-AP-LP antibody, anti-DNP-LP antibody, or anti-fluorophore-LP antibody, respectively.
  • the above antibodies are produced by animal species A, such as goat. This first antibody is then recognized by second antibody against species A.
  • the second antibody is conjugated with HRP, Polymer-HRP, AP, Polymer-AP, or fluorophore and is capable of binding to one or more first antibodies.
  • HRP or AP can be detected visually by chromogenic dyes such as Diaminobenzidine (DAB) or Fast Red.
  • DAB Diaminobenzidine
  • the result here is further signal amplification in addition to the RNAscope signal amplification.
  • the TSA-based ISH signal amplification method was used in combination with RNAscope®.
  • the RNAscope comprises a set of two or more capture probes, a plurality of label probes, a plurality of amplifiers, and a plurality of preamplifiers.
  • the target nucleic acid is hybridized to the set of capture probes.
  • the set of capture probes is then hybridized to a preamplifier.
  • the preamplifier is further hybridized to a plurality of amplifiers.
  • Each amplifier is then hybridized to a plurality of label probes.
  • Each label probe of the RNAscope® complex structure is conjugated to an HRP molecule forming an HRP-LP complex.
  • the HRP-LP complex is then bind to a plurality of Tyramide-Biotin or Fluorophore-Biotin molecules.
  • the HRP-LP complex catalyze multiple copies of Tyramide-Biotin or Fluorophore-Biotin to covalently bind to electron-rich moieties of proteins in the vicinity of LP-HRP binding sites.
  • the Tyramide-Biotin or Fluorophore-Biotin is then subsequently detected by detection labels.
  • the detection labels are a combination of avidin/streptavidin-HRP and chromogenic substrate or a combination of avidin/streptavidin-AP and chromogenic substrate.
  • the HRP or AP on the avidin/streptavidin-HRP or avidin/streptavidin-AP complexes are detected visually by chromogenic substrates such as DAB or Fast Red. Since many copies of Tyramide-Biotin or Fluorophore-Biotin can be deposited around each LP-HRP binding site, the intensity of end signal, which is the sum of RNAscope amplification and TSA amplification, is significantly increased.
  • tyramide in other embodiments of the TSA-based ISH signal amplification method, can also be conjugated to other haptens such as digoxigenin (DIG), trinitrophenyl (TNP), Dinotrophenyl (DNP) and fluorescent dyes.
  • DIG digoxigenin
  • TNP trinitrophenyl
  • DNP Dinotrophenyl
  • fluorescent dyes when fluorescent dye molecules are used in tyramide conjugation, the tyramide-dye conjugates can be directly used in RNAscope-TSA system for simple and effective fluorescent (FISH) detection.
  • RNAscope® with general ISH signal amplification methods disclosed in this invention can be further fine-tuned to increase the sensitivity and specificity of signal amplification.
  • RNAscope® + general ISH signal amplification approach RNAscope®'s major contribution is assay specificity.
  • the RNAscope® also provides modest signal amplification of the nucleic acid target with high specificity.
  • One of the general ISH signal amplification methods' contributions is their signal boosting power.
  • the general ISH signal amplification methods further amplify the target nucleic acid signal to a much higher level.
  • RNAscope® with general ISH signal amplification methods faces the problem of elevated background noise in signal detection.
  • the present invention discloses an intelligent way to reduce the level of false positive signals.
  • RNAscope® One balancing factor for the combination of RNAscope® with general ISH signal amplification methods disclosed in this invention is the size of the preamplifier used in RNAscope®.
  • the preamplifier of the RNAscope® could be a very large molecule if it is designed to bind to a large number of amplifiers.
  • the preamplifier may have the capacity to hybridize to more than 20 amplifiers.
  • Such a large molecule is prone to bind nonspecifically in cellular matrix during in situ hybridization.
  • one of the preamplifiers binds to sample nonspecifically, it will attract the same large number of amplifiers and label probes onto itself as it does when binding to a target nucleic acid.
  • RNAscope® In the case when RNAscope® is used alone, the number of label probes which are bound to the few nonspecific binding preamplifiers may not generate any observable signals. However, if the signal of these label probes is further boosted by the additional signal amplification by a subsequent general ISH signal amplification, the false positive signal will be high enough to be detected.
  • one preamplifier is designed to bind to between 1 to 16 amplifiers. In a more preferred embodiment, one preamplifier is designed to bind to between 2 to 10 amplifiers. In another preferred embodiment, one preamplifier is designed to bind to between 2 to 5 amplifiers.
  • RNAscope® By balancing the amplification powers and specificity of RNAscope® and TSA in the assay, an optimum signal-to-noise ratio has been achieved for in situ RNA detection in routine clinical tissue specimen (see example 1 and Fig. 5 ).
  • RNAscope® A powerful in situ hybridization method called RNAscope® was recently developed by Advanced Cell Diagnostics, Inc. (United States Patent No. 7,709,198 , incorporated herein by reference in its entirety). Since RNAscope® is part of the method disclosed in the present invention, the working principle of RNAscope® will be described below for better understanding of the invention.
  • RNAscope® allows for direct visualization of RNA in situ. This method utilizes the oligonucleotide probe sets and novel signal amplification systems described below.
  • the assay can be used on a variety of sample types including cultured cells, peripheral blood mononuclear cells (PBMCs), frozen tissue, and formalin-fixed paraffin embedded (FFPE) tissue.
  • PBMCs peripheral blood mononuclear cells
  • FFPE formalin-fixed paraffin embedded
  • the assay can utilize both chromogenic and fluorescent detection reagents.
  • the RNAscope® assay technology provides multiplex nucleic acid assays in single cells (see Fig. 1 ).
  • At the core of this technology is the "double Z" probe design, which allows robust amplification of specific hybridization signals without also amplifying nonspecific events.
  • Each capture probe (“Z”) has a target-specific sequence which binds to the target mRNA, a spacer, and a "tail” sequence.
  • Two capture probes (double Z) hybridize contiguously onto a target mRNA, and the two "tail” sequences form a 28-base hybridization site for the preamplifier.
  • the double Z probe design ensures high fidelity of signal amplification because: 1) it is highly unlikely that a pair of target probes will hybridize nonspecifically juxtaposed to each other to form a binding site for the preamplifier; and 2) neither tail alone can bind efficiently to the preamplifier under the assay conditions.
  • the preamplifier, amplifier and label probe are hybridized sequentially to each capture probe pair, resulting in the accumulation of as many as 8,000 label molecules per 1 kb of target RNA.
  • the label probe can be conjugated to either a fluorophore or a chromogenic enzyme (e.g. , HRP or AP), enabling viewing of hybridization signals under a standard bright-field or epifluorescent microscope, respectively. With a fluorescent label probe, the signals can contain at least 100-fold more fluorescent molecules than traditional RNA fluorescent ISH methods and are readily visible under a standard fluorescent microscope.
  • RNAscope® was used to detect a target nucleic acid.
  • a sample comprising one or more cell is provided.
  • the cell tested comprises, or is suspected of comprising, the target nucleic acid.
  • Provided in the assay are: a set of capture probes comprising two or more capture probes, a label probe comprising a label, and optionally preamplifiers and amplifiers.
  • the set of capture probes is hybridized, in the cell, to the target nucleic acid.
  • the label probe is captured to the set of capture probes, thereby capturing the label probe to the target nucleic acid.
  • the signal from the label is then detected. Since the label is associated with the target nucleic acid through the capture probes, presence of the label(s) in the cell indicates the presence of the corresponding nucleic acid target(s) in the cell.
  • the methods are optionally quantitative.
  • an intensity of the signal can be measured, and the intensity of the signal can be correlated with a quantity of the target nucleic acid in the cell.
  • a signal spot can be counted for each copy of the target nucleic acid in order to quantify them.
  • the label probes bind directly to the capture probes.
  • the label probes are captured to the capture probes indirectly, for example, through binding of preamplifiers and/or amplifiers.
  • Use of amplifiers and preamplifiers can be advantageous in increasing signal strength, since they can facilitate binding of large numbers of label probes to each nucleic acid target.
  • the indirect capture approach is preferred because it enables the label probe to be target-independent and further disclosure will show that it can offer better specificity and sensitivity.
  • the capture probe is specially designed in the RNAscope® assay.
  • each capture probe there is at least one section, T, complementary to a section on the target molecule, and another section, L, complementary to a section on the label probe.
  • the T and L sections are connected by a section C.
  • two adjacent capture probes are incorporated in a probe set targeting a gene of interest.
  • T1 and T2 are designed to be complementary to two unique and adjacent sections on the target nucleic acid.
  • L1 and L2 which can be different or the same, are complementary to two adjacent sections on the label probe.
  • Their binding sections, T, L or both are designed so that the linkage between the label probe and the target is unstable and tends to fall off at hybridization temperature when only one of the capture probes is in place.
  • the melting temperature, T m of the T sections of the two capture probes are designed to be significantly above the hybridization temperature while the T m of the L sections is below the hybridization temperature.
  • the T sections can be 20-30 nucleotides in length while the L sections are 13-15 nucleotides in length; C can be 0 to 10 nucleotides in length, e.g ., 5 nucleotides.
  • T m of the T sections is below hybridization temperature while T m of the L sections is substantially above.
  • the linkage between the label probe and the target can only survive the hybridization when both capture probes are hybridized to the target in a cooperative fashion. See U.S. Patent No. 7,709,198 for additional details on design of capture probes.
  • the capture probes preferably hybridize to nonoverlapping polynucleotide sequences in their respective nucleic acid target.
  • the capture probes can, but need not, cover a contiguous region of the nucleic acid target.
  • Blocking probes, polynucleotides which hybridize to regions of the nucleic acid target not occupied by target probes, are optionally provided and hybridized to the target.
  • the corresponding capture probes and blocking probes are preferably complementary to physically distinct, nonoverlapping sequences in the nucleic acid target, which nonoverlapping sequences are preferably, but not necessarily, contiguous. Having the capture probes and optional blocking probes be contiguous with each other can in some embodiments enhance hybridization strength, remove secondary structure, and ensure more consistent and reproducible signal.
  • the cell In detection of nucleic acid targets in a cell, the cell is typically fixed and permeabilized before hybridization of the capture probes, to retain the nucleic acid targets in the cell and to permit the capture probes, label probes, etc. to enter the cell.
  • the cell is optionally washed to remove materials not captured to one of the nucleic acid targets.
  • the cell can be washed after any of various steps, for example, after hybridization of the capture probes to the nucleic acid targets to remove unbound capture probes, after hybridization of the preamplifiers, amplifiers, and/or label probes to the capture probes, and/or the like.
  • the cell is in suspension for all or most of the steps of the method, for ease of handling.
  • the methods are also applicable to cells in solid tissue samples (e.g. , tissue sections) and/or cells immobilized on a substrate ( e.g. , a slide or other surface).
  • the cell is in suspension in the sample comprising the cell, and/or the cell is in suspension during the hybridizing, capturing, and/or detecting steps.
  • the cell can be in suspension in the sample and during the hybridization, capture, optional washing, and detection steps.
  • the cell is in suspension in the sample comprising the cell, and the cell is fixed on a substrate during the hybridizing, capturing, and/or detecting steps.
  • the cell can be in suspension during the hybridization, capture, and optional washing steps and immobilized on a substrate during the detection step.
  • the sample comprises a tissue section.
  • one aspect of the invention provides multiplex nucleic acid assays.
  • one general class of embodiments includes methods of detecting two or more target nucleic acids.
  • method for detecting a single target nucleic acid using RNAscope® has been described.
  • method for detecting a single target nucleic acid using a combination of RNAscope® and general ISH signal amplification method will be described first.
  • Method for detecting two or more target nucleic acids using a combination of RNAscope® and general ISH signal amplification method will be described next.
  • the detection of two or more target nucleic acids can be conducted in a same way as that of detecting a single target nucleic acids except for using different, target-specific, labels and probes.
  • a method of detecting a single target nucleic acid is provided.
  • a sample comprising or suspected of comprising the target nucleic acid is provided.
  • a set of two or more capture probes wherein the capture probes are capable of binding to the target nucleic acid, signal generating multimers, and signal amplification probes.
  • the signal generating multimer comprises at least a label probe.
  • the signal generating multimer comprises a label probe and an amplifier.
  • the amplifier is capable of hybridizing to the label probe, and is also capable of hybridizing to the set of two or more capture probes.
  • the signal generating multimer comprises a label probe, an amplifier capable of hybridizing to the label, and a preamplifier capable of hybridizing to the amplifier and also capable of hybridizing to the set of two or more capture probes.
  • the signal amplification probes are made of components used in general ISH signal amplification assays.
  • the signal amplification probe comprises: a biotin molecule which is capable of conjugating to the label probe of RNAscope®, an avidin/streptavidin molecule which is capable of binding to the biotin molecule, and additional biotin molecules being conjugated to HRP, AP, or fluorophore and are capable of binding to the avidin/streptavidin molecule.
  • the signal amplification probe comprises: a HRP, DNP, fluorophore, or AP molecule which is capable of conjugating to said label probe, one or more first antibody which is capable of binding to the HRP, DNP, fluorophore, or AP molecule, and one or more second antibody which is conjugated with HRP, Polymer-HRP, AP, or Polymer-AP and is capable of binding to one or more of the first antibody.
  • the signal amplification probe comprises: a HRP molecule which is capable of conjugating to said label probe, a plurality of tyramide-biotin or tyramide-fluorophore molecules which are capable of reacting with the HRP molecule, and detection labels which are capable binding to the tyramide-biotin or tyramide-fluorophore molecules and thus display the target nucleic acid visually.
  • the detection labels are a combination of acivdin/strepavidin-HRP and a chromogenic substrate or a combination of acivdin/strepavidin-HRP and a chromogenic substrate.
  • a sample comprising or suspected of comprising the target nucleic acid is first provided.
  • the target nucleic acid is then hybridized to the set of two or more capture probes.
  • the signal generating multimer is then added and being hybridized to the set of two or more capture probes and thereby being captured to the target nucleic acid.
  • Components of the signal amplification probe are then added to the assay solution sequentially and bind to the signal generating multimer and thereby being captured to the target nucleic acid.
  • the labels in the signal amplification probe are then detected by flow cytometer or other visualization devices. The presence, absence, or amount of the labels allows a sensitive detection and localization of the target nucleic acid in the sample.
  • a sample comprising or suspected of comprising the two target nucleic acids is provided. Also provided are two or more sets of capture probes, each set comprising two or more capture probes. The first set of capture probe is capable of hybridizing to the first target nucleic acid but not the second target nucleic acid, and the second set of capture probe is capable of hybridizing to the second target nucleic acid but not the first target nucleic acid. Further provided are a first signal generating multimer and a first signal amplification probe, together with a second signal generating multimer and a second signal amplification probe.
  • the first signal generating multimer can hybridize to the first set of capture probe but not the second set of capture probe.
  • the second signal generating multimer can hybridize to the second set of capture probe but not the first set of capture probe.
  • the first signal generating multimer has a first label probe that hybridize, either directly or indirectly through preamplifiers and amplifiers, to the first set of capture probe.
  • the second signal generating multimer has a second label probe that hybridize, either directly or indirectly through preamplifiers and amplifiers, to the second set of capture probe.
  • the first label probe is different from the second label probe.
  • the first signal generating multimer and the second signal generating multimer are labeled with different signal amplification probes sequentially.
  • the two signal amplification probes can be a dual-color CISH or a dual-color FISH.
  • the first signal generating multimer is labeled with the first signal amplification probe.
  • the first signal amplification probe is a TSA-based ISH signal amplification which linked with the label probe of RNAscope® amplification via a conjugated LP-HRP complex.
  • the label probe of RNAscope® is first conjugated with HRP.
  • the conjugated LP-HRP complex is then detected with sequential incubation of tyramide-biotin, streptavidin-HRP and color substrate DAB, resulting in brown signal for the first target nucleic acid.
  • the residual labeling agents for the first signal generating multimer are blocked in order to prevent the labels from reacting with the second signal generating multimer which is going to be added in the third step.
  • the second signal generating multimer is labeled with the second signal amplification probe.
  • the label probe of RNAscope® is first conjugated with Dinotrophenyl (DNP).
  • DNP Dinotrophenyl
  • the conjugated LP-DNP complex is then detected with sequential incubation of anti-DNP-AP and color substrate Fast Red, resulting in red signal for the first target nucleic acid.
  • the above methods are also useful for multiplex detection of nucleic acids, including sequential detection of more than three nucleic acid targets.
  • the cell optionally comprises or is suspected of comprising a third, fourth, fifth, sixth, seventh or even more nucleic acid target can be analyzed in the similar way described above.
  • kits for detecting one or more target nucleic acid includes a target nucleic acid, an RNAscope® component and a general ISH signal amplification component.
  • the kit includes a target nucleic acid, sets of capture probes capable of hybridizing to the target nucleic acid, a signal generating multimer capable of hybridizing to set of capture probes, and a signal amplification probe capable of binding to the label probe.
  • the signal generating multimer comprises a label probe and the signal amplification probe comprises a label.
  • the signal generating multimer in the kit can include appropriate amplifiers, preamplifiers.
  • the kit may also include a solid support.
  • the ISH signal amplification component of the kit can be any ISH signal amplification system.
  • it can be the three ISH signal amplification systems described in Section 1.1 of the present invention.
  • the kit can be used for detection of any nucleic acids described earlier in this application, including, not not limited to DNA, cDNA, RNA, and mRNA.
  • kits many components can be represented with two or more different subsets.
  • the label probes can include two subsets, hybridizing to the first and the second target nucleic acids, respectively.
  • the signal generating multimer can include two different label probe, each hybridizes, either directly or indirectly, to the first and the second capture probes.
  • the kit can also include two different signal amplification probes, each conjugates to the first and second label probes. The different signal amplification probes are distinguishable from each other by binding to different color substrates.
  • the kit includes a blocking agent which is capable of blocking the residual labeling agents for the first signal generating multimer and thus ensuring the specific binding of the second signal generating multimer to the second target nucleic acid.
  • RNAscope® shows how a combination of RNAscope® and general ISH signal amplification methods further enhanced by an optimized preamplifier/amplifier ratio achieves a significantly improved specificity and sensitivity for in situ RNA detection.
  • the basic assay procedure can be done within a day and generally includes the following steps.
  • a sample containing a plurality of cells suspected of comprising a target nucleic acid HPRT1 is provided.
  • cells either fixed on a solid support or in suspension are hybridized to the following series of oligonucleotide probes.
  • a set of capture probes is hybridized to the target RNA inside the cells.
  • preamplifier molecules are hybridized to the capture probes, providing a bridge for the hybridization of amplifier molecules.
  • Multiple amplifier molecules are further hybridized to a preamplifier, e.g ., up to 20 amplifiers to each preamplifier.
  • Multiple label probes are then hybridized to an amplifier, e.g . up to 20 label probes to each amplifier.
  • the signal intensity is enhanced further by TSA-based ISH signal amplification.
  • An HRP is conjugated to each label probe.
  • Reagent containing tyramide-biotin molecules is then added to the assay solution.
  • the HRP-conjugated-label-probe catalyzes multiple copies of tyramide-biotin to covalently bind to the electron-rich moieties of proteins in the vicinity of the HRP-label probe binding site.
  • the tyramide-biotin molecule is then detected by avidin/streptavidin-HRP, where avidin/streptavidin-HRP can be visualized by label substrate DAB which shows brown color.
  • RNAscope® + TSA-based ISH signal amplification method signals obtained from RNAscope® + TSA-based ISH signal amplification method was significantly stronger than that obtained from RNAscope® alone.
  • Nonspecific binding was prevented or minimized by designing probe sets targeting a specific mRNA sequence using a double "Z" probe design.
  • the double "Z" capture probes were designed based on the sequence of the HPRT1 and were prescreened against the GenBank database to ensure minimal cross-hybridization with unintended nucleic acid sequences.
  • two neighboring probes each contain a target-hybridizing sequence, e.g ., 20 to 30 base in length with a T m significantly above the assay temperature, and a preamplifier-hybridizing sequence, e.g. , only 14 bases in length with a T m well below the assay temperature.
  • a single capture probe is able to bind to target RNA strongly and stably during hybridization, but will bind to the preamplifier weakly and unstably due to the 14 base pair region of homology having a T m well below the assay temperature.
  • the combined hybridization strength e.g ., of 28 complementary base pairs, holds the preamplifier strongly and stably at the assay temperature, enabling signal amplification to occur.
  • Such a double "Z" design ensures high detection specificity and simplifies probe design for simultaneous detection of multiple targets.
  • the signal boosting power of the TSA-based ISH signal amplification method Due to the signal boosting power of the TSA-based ISH signal amplification method, the nonspecific binding of preamplifier in cellular matrix during in situ hybridization could be amplified and consequently produces a false positive signal. This problem becomes serious when the preamplifier is designed to bind large number of amplifiers and thus is large in size. Power of amplification is gained at the cost of lost binding specificity. We solved this problem by moderately reducing the number of amplifiers a preamplifier is designed to bind to. The end result is increased the signal-to-noise ratio. The lost amplification power is compensated by signal boosting power of the ISH signal amplification methods, particularly the TSA-based ISH signal amplification method.
  • Preamplifier PREAMP1 is designed to bind to 20 amplifiers.
  • Preamplifier PREAMP2 is designed to bind to 16 amplifiers.
  • Preamplifier PREAMP3 is designed to bind to 10 amplifiers.
  • Preamplifier PREAMP4 is designed to bind to 5 amplifiers.
  • PREAMP2 has improved signal-to-noise ratio over PREAMP 1.
  • PREAMP3 and PREAMP4 have the best signal-to-noise ratio among all the PREAMPs tested (data not shown).
  • 18S gene is captured on capture probes designed for 18S.
  • the capture probe for 18S is designed as described in example 1.
  • the capture-probe-hybridized-18S gene is then sequentially hybridized to preamplifiers, amplifiers and label probes.
  • the label probe is then conjugated to HRP.
  • the conjugated LP-HRP complexes are then detected with sequential incubations of tyramide-biotin, streptavidin-HRP and DAB. The product of the hybridization produces brown color at locations where target gene 18Ss are detected.
  • Her-2 gene is captured on capture probes designed for Her-2.
  • the capture probe for Her-2 is designed as described in example 1.
  • the capture-probe-hybridized-Her-2 gene is then sequentially hybridized to preamplifiers, amplifiers and label probes.
  • the label probe is then conjugated to Diotrophenyl (DNP).
  • DNP Diotrophenyl
  • the conjugated LP-DNPs are then detected with incubation of anti-DNP-AP.
  • the alkaline phosphatase (AP) molecule on the anti-DNP-AP complex is then detected by dye reagent Fast Red.
  • the product of the hybridization produces red signal for target gene Her-2.
  • the method disclosed in this invention can be used to detect two RNA transcripts.
  • the relative intensity of the signals can be used to compare gene expression levels of the two genes.
  • the invention refers inter alia to the following items:

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
EP15194058.2A 2010-10-21 2011-10-21 Procédé ultrasensible de détection in situ d'acides nucléiques Active EP3034625B1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US40550310P 2010-10-21 2010-10-21
US41227610P 2010-11-10 2010-11-10
EP11785174.1A EP2630260B1 (fr) 2010-10-21 2011-10-21 Procédé ultrasensible de détection in situ d'acides nucléiques

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
EP11785174.1A Division EP2630260B1 (fr) 2010-10-21 2011-10-21 Procédé ultrasensible de détection in situ d'acides nucléiques

Publications (2)

Publication Number Publication Date
EP3034625A1 true EP3034625A1 (fr) 2016-06-22
EP3034625B1 EP3034625B1 (fr) 2017-10-04

Family

ID=44999880

Family Applications (2)

Application Number Title Priority Date Filing Date
EP11785174.1A Active EP2630260B1 (fr) 2010-10-21 2011-10-21 Procédé ultrasensible de détection in situ d'acides nucléiques
EP15194058.2A Active EP3034625B1 (fr) 2010-10-21 2011-10-21 Procédé ultrasensible de détection in situ d'acides nucléiques

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP11785174.1A Active EP2630260B1 (fr) 2010-10-21 2011-10-21 Procédé ultrasensible de détection in situ d'acides nucléiques

Country Status (5)

Country Link
US (3) US8658361B2 (fr)
EP (2) EP2630260B1 (fr)
CN (3) CN104849472A (fr)
ES (2) ES2562821T3 (fr)
WO (1) WO2012054795A1 (fr)

Families Citing this family (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090081688A1 (en) * 2005-06-20 2009-03-26 Advanced Cell Diagnostics Methods of detecting nucleic acids in individual cells and of identifying rare cells from large heterogeneous cell populations
ES2565670T3 (es) 2005-06-20 2016-04-06 Advanced Cell Diagnostics, Inc. Métodos de detección de ácidos nucleicos en células individuales y de identificación de células raras en grandes poblaciones de células heterogéneas
US20130023433A1 (en) * 2009-09-28 2013-01-24 Yuling Luo Methods of detecting nucleic acid sequences with high specificity
CA2787487C (fr) * 2010-02-26 2018-07-24 Ventana Medical Systems, Inc. Sondes polytag
US10787701B2 (en) 2010-04-05 2020-09-29 Prognosys Biosciences, Inc. Spatially encoded biological assays
US20190300945A1 (en) 2010-04-05 2019-10-03 Prognosys Biosciences, Inc. Spatially Encoded Biological Assays
WO2012048113A2 (fr) 2010-10-07 2012-04-12 The General Hospital Corporation Biomarqueurs de cancer
CN104849472A (zh) 2010-10-21 2015-08-19 领先细胞医疗诊断有限公司 用于原位检测核酸的超灵敏方法
GB201106254D0 (en) 2011-04-13 2011-05-25 Frisen Jonas Method and product
WO2013055995A2 (fr) 2011-10-14 2013-04-18 President And Fellows Of Harvard College Séquençage par assemblage structurel
ES2953308T3 (es) 2011-12-22 2023-11-10 Harvard College Composiciones y métodos para la detección de analitos
US11021737B2 (en) 2011-12-22 2021-06-01 President And Fellows Of Harvard College Compositions and methods for analyte detection
WO2013148498A1 (fr) 2012-03-27 2013-10-03 Ventana Medical Systems, Inc. Conjugués de signalisation, et procédés d'utilisation
US20140178869A1 (en) 2012-04-05 2014-06-26 Advanced Cell Diagnostics, Inc. Detection of immunoglobulin light chain restriction by rna in situ hybridization
WO2013184754A2 (fr) 2012-06-05 2013-12-12 President And Fellows Of Harvard College Séquençage spatial d'acides nucléiques à l'aide de sondes d'origami d'adn
EP3578666A1 (fr) 2013-03-12 2019-12-11 President and Fellows of Harvard College Procédé de génération d'une matrice contenant un acide nucléique tridimensionnel
CA2914816A1 (fr) * 2013-06-14 2014-12-18 Nanostring Technologies, Inc. Systeme rapporteur multiplexable a base d'etiquettes
CN105849275B (zh) 2013-06-25 2020-03-17 普罗格诺西斯生物科学公司 检测样品中生物靶标的空间分布的方法和系统
US20150247204A1 (en) 2014-02-07 2015-09-03 The General Hopital Corporation Differential diagnosis of hepatic neoplasms
WO2015123565A1 (fr) 2014-02-14 2015-08-20 The General Hospital Corporation Méthodes de diagnostic d'une maladie liée à l'igg4
EP3617322A1 (fr) * 2014-02-24 2020-03-04 Ventana Medical Systems, Inc. Détection automatisée d'arn à l'aide de sondes oligonucléotidiques d'arn 2'-o-méthyle marquées et de systèmes d'amplification de signal
US20150247205A1 (en) 2014-02-28 2015-09-03 The General Hospital Corporation Diagnosis of multiple myeloma and lymphoma
US20160063415A1 (en) * 2014-06-18 2016-03-03 North Central Cooperative Crop production system
EP3760208B1 (fr) 2014-06-25 2024-05-29 The General Hospital Corporation Ciblage de hsatii (human satellite ii)
EP3175023A4 (fr) 2014-07-30 2018-03-07 President and Fellows of Harvard College Systèmes et méthodes permettant de déterminer des acides nucléiques
US20160046984A1 (en) * 2014-08-15 2016-02-18 Affymetrix, Inc. Robust Detection of Nucleic Acids in Situ
US11124822B2 (en) * 2014-10-17 2021-09-21 Carnegie Mellon University Enhanced biomolecule detection assays based on tyramide signal amplification and gammaPNA probes
US9851330B2 (en) * 2015-03-20 2017-12-26 Konica Minolta Laboratory U.S.A., Inc. Rapid, highly-sensitive, and highly-specific nucleic acid detection
EP3530752B1 (fr) 2015-04-10 2021-03-24 Spatial Transcriptomics AB Analyse de plusieurs acides nucléiques spatialement différenciés de spécimens biologiques
US20180171382A1 (en) * 2015-06-26 2018-06-21 Albert-Ludwigs-Universität Freiburg Branched proximity hybridization assay
ES2893100T3 (es) 2015-10-12 2022-02-08 Advanced Cell Diagnostics Inc Detección in situ de variantes de nucleótidos en muestras con un elevado nivel de ruido, y composiciones y métodos relacionados con ésta
WO2017079406A1 (fr) 2015-11-03 2017-05-11 President And Fellows Of Harvard College Procédé et appareil pour imagerie volumétrique d'une matrice tridimensionnelle contenant des acides nucléiques
CN105861669B (zh) * 2016-04-22 2019-10-18 郑州科蒂亚生物技术有限公司 一种快速捕获杂交靶标物分支链dna信号放大的检测方法
CA3210120C (fr) 2016-04-25 2024-04-09 President And Fellows Of Harvard College Procedes de reaction en chaine d'hybridation pour la detection moleculaire in situ
EP3472359B1 (fr) 2016-06-21 2022-03-16 10X Genomics, Inc. Séquençage d'acide nucléique
EP3481843B1 (fr) 2016-07-05 2022-01-26 California Institute of Technology Réaction en chaîne d'hybridation d'initiateurs fractionnaire
US11118212B2 (en) 2016-07-29 2021-09-14 University Of Utah Research Foundation Methods for detecting rapidly processed introns to evaluate allelic expression
WO2018045186A1 (fr) 2016-08-31 2018-03-08 President And Fellows Of Harvard College Procédés de combinaison de la détection de biomolécules dans un dosage unique à l'aide d'un séquençage fluorescent in situ
CN110225980B (zh) 2016-11-21 2023-01-06 纳米线科技公司 化学组合物及其使用方法
DK3555313T3 (da) 2016-12-16 2021-08-09 Aratome Llc Molekylær detektering ved hjælp af ligeringsamplifikation
US10280445B2 (en) 2017-03-09 2019-05-07 Diagnostic Biosystems Chromogen layering for color generation
WO2018218150A1 (fr) 2017-05-26 2018-11-29 President And Fellows Of Harvard College Systèmes et méthodes de criblage à base d'image à haut débit
US10379015B2 (en) * 2017-06-04 2019-08-13 Diagnostic Biosystems Method for labeling concentration density differentials of an analyte in a biological sample
CN107227350A (zh) * 2017-06-09 2017-10-03 苏州达麦迪生物医学科技有限公司 一种快速检测alk基因断裂的探针组、试剂盒及方法
CN111263819A (zh) 2017-10-06 2020-06-09 卡特阿纳公司 Rna模板化连接
CN107764804B (zh) * 2017-10-09 2020-02-07 郑州大学 一种自组装聚hrp探针、制备方法、试剂盒及其应用
EP4317972A3 (fr) 2018-02-06 2024-03-13 The General Hospital Corporation Arn à motifs répétés utilisés comme biomarqueurs de réponse immunitaire tumorale
CN108504777A (zh) * 2018-04-03 2018-09-07 新乡医学院第三附属医院 一种基于信号放大检测hcv病毒载量的方法
WO2019199643A1 (fr) 2018-04-09 2019-10-17 Bio-Techne Corporation Procédés d'amélioration supplémentaire de l'amplification de signal pour la détection in situ d'acides nucléiques
CA3099909A1 (fr) 2018-05-14 2019-11-21 Nanostring Technologies, Inc. Compositions chimiques et leurs methodes d'utilisation
CN108949913B (zh) * 2018-07-31 2021-05-25 四川大学华西医院 包含通用荧光消减探针核酸分子的基因检测试剂盒及其应用
AU2019398322A1 (en) * 2018-12-13 2021-06-24 President And Fellows Of Harvard College Amplification methods and systems for MERFISH and other applications
WO2020168162A1 (fr) 2019-02-15 2020-08-20 Bio-Techne Corporation Procédés de détection multiplexe d'acides nucléiques par hybridation in situ
EP3976817A1 (fr) 2019-05-31 2022-04-06 10X Genomics, Inc. Procédé de détection de molécules d'acide nucléique cible
CN112113820A (zh) * 2020-05-28 2020-12-22 王剑 一种细胞病理学样本的染色制片方法
US11459567B2 (en) 2020-06-24 2022-10-04 Patricia Virginia Elizalde Specific siRNA molecules, composition and use thereof for the treatment of triple negative breast cancer
CN114544966B (zh) * 2020-11-11 2023-05-09 艾克发(北京)生物技术有限公司 一种多重信号放大系统及其在免疫吸附夹心法检测中的应用
EP4284925A1 (fr) 2021-01-26 2023-12-06 California Institute of Technology Arn guides conditionnels allostériques pour la régulation sélective de cellules de crispr/cas
JP7062803B1 (ja) 2021-03-31 2022-05-06 積水メディカル株式会社 抗薬物抗体測定方法
WO2022250774A1 (fr) * 2021-05-24 2022-12-01 California Institute Of Technology Amplification liée attachée à une radiance exponentielle
EP4359798A1 (fr) 2021-06-23 2024-05-01 Janssen Biotech, Inc. Utilisation d'elisa unicellulaire à partir de cellules déparaffinées pour la détection de molécules d'intérêt
US20230279475A1 (en) 2022-01-21 2023-09-07 10X Genomics, Inc. Multiple readout signals for analyzing a sample
WO2023192616A1 (fr) 2022-04-01 2023-10-05 10X Genomics, Inc. Compositions et procédés de masquage ciblé d'autofluorescence
WO2023196526A1 (fr) 2022-04-06 2023-10-12 10X Genomics, Inc. Procédés d'analyse multiplex de cellules
US20240035071A1 (en) 2022-06-17 2024-02-01 10X Genomics, Inc. Catalytic de-crosslinking of samples for in situ analysis
WO2024081869A1 (fr) 2022-10-14 2024-04-18 10X Genomics, Inc. Procédés d'analyse d'échantillons biologiques

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
EP0795610A1 (fr) * 1996-03-13 1997-09-17 Becton, Dickinson and Company Amplification de signal d'hybrisation "in situ" par dépÔt de tyramine biotinilée
WO2007002006A2 (fr) * 2005-06-20 2007-01-04 Panomics, Inc. Detection multiplex d’acides nucleiques

Family Cites Families (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4994373A (en) 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
US4888278A (en) 1985-10-22 1989-12-19 University Of Massachusetts Medical Center In-situ hybridization to detect nucleic acid sequences in morphologically intact cells
US5985549A (en) 1985-10-22 1999-11-16 University Of Massachusetts Non-isotopic in-situ hybridization method for detection of nucleic acids
US5093232A (en) 1985-12-11 1992-03-03 Chiron Corporation Nucleic acid probes
US4868105A (en) 1985-12-11 1989-09-19 Chiron Corporation Solution phase nucleic acid sandwich assay
US5122599A (en) 1986-08-13 1992-06-16 Molecular Diagnostics, Inc. CDNAS coding for members of the carcinoembryonic antigen family
US5359100A (en) 1987-10-15 1994-10-25 Chiron Corporation Bifunctional blocked phosphoramidites useful in making nucleic acid mutimers
US5124246A (en) 1987-10-15 1992-06-23 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using same
SE8801537D0 (sv) 1988-04-26 1988-04-26 Ellco Food Ab Cellodlingsmedium samt forfarande for dess framstellning
US5374524A (en) 1988-05-10 1994-12-20 E. I. Du Pont De Nemours And Company Solution sandwich hybridization, capture and detection of amplified nucleic acids
US6242184B1 (en) 1988-10-18 2001-06-05 University Of Massachusetts In-situ hybridization of single-copy and multiple-copy nucleic acid sequences
CA1339729C (fr) 1988-10-26 1998-03-17 Wayne D. Lancaster Sequences d'and pour virus des papillomes humains de type 52 methode d'utilisation
US5334499A (en) 1989-04-17 1994-08-02 Eastman Kodak Company Methods of extracting, amplifying and detecting a nucleic acid from whole blood or PBMC fraction
US5231015A (en) 1989-10-18 1993-07-27 Eastman Kodak Company Methods of extracting nucleic acids and pcr amplification without using a proteolytic enzyme
US5185244A (en) 1989-12-08 1993-02-09 Emory University Genetic test for hereditary neuromuscular disease
US5130423A (en) 1990-07-13 1992-07-14 Microprobe Corporation Non-corrosive compositions and methods useful for the extraction of nucleic acids
US5849481A (en) 1990-07-27 1998-12-15 Chiron Corporation Nucleic acid hybridization assays employing large comb-type branched polynucleotides
US5747244A (en) 1991-12-23 1998-05-05 Chiron Corporation Nucleic acid probes immobilized on polystyrene surfaces
WO1994000598A1 (fr) 1992-06-19 1994-01-06 Northwestern University Procede de detection de sequences d'acide nucleique amplifiees de cellules par cytometrie en flux
SE9201929D0 (sv) 1992-06-23 1992-06-23 Pharmacia Lkb Biotech Method and system for molecular-biological diagnostics
US5633134A (en) 1992-10-06 1997-05-27 Ig Laboratories, Inc. Method for simultaneously detecting multiple mutations in a DNA sample
US5386024A (en) 1993-02-10 1995-01-31 Gen-Probe Incorporated Method to prepare nucleic acids from a biological sample using low pH and acid protease
WO1995003428A1 (fr) 1993-07-20 1995-02-02 University Of Massachusetts Medical Center Procede d'hybridation in vivo d'acide nucleique
US5681697A (en) * 1993-12-08 1997-10-28 Chiron Corporation Solution phase nucleic acid sandwich assays having reduced background noise and kits therefor
US5523204A (en) 1993-12-10 1996-06-04 Becton Dickinson And Company Detection of nucleic acids in cells by strand displacement amplification
US5681702A (en) 1994-08-30 1997-10-28 Chiron Corporation Reduction of nonspecific hybridization by using novel base-pairing schemes
US5780227A (en) 1995-06-07 1998-07-14 Sheridan; Patrick J. Oligonucleotide probe conjugated to a purified hydrophilic alkaline phosphatase and uses thereof
US5945515A (en) 1995-07-31 1999-08-31 Chomczynski; Piotr Product and process for isolating DNA, RNA and proteins
US5804684A (en) 1995-08-24 1998-09-08 The Theobald Smith Research Institute, Inc. Method for isolating nucleic acids
DE19537952A1 (de) * 1995-10-12 1997-04-17 Boehringer Mannheim Gmbh Verfahren zum Nachweis eines Analyten
US5866331A (en) 1995-10-20 1999-02-02 University Of Massachusetts Single molecule detection by in situ hybridization
US6418382B2 (en) 1995-10-24 2002-07-09 Curagen Corporation Method and apparatus for identifying, classifying, or quantifying DNA sequences in a sample without sequencing
US6007994A (en) 1995-12-22 1999-12-28 Yale University Multiparametric fluorescence in situ hybridization
US6750016B2 (en) 1996-07-29 2004-06-15 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
US20020172953A1 (en) 1996-07-29 2002-11-21 Mirkin Chad A. Movement of biomolecule-coated nanoparticles in an electric field
US6352827B1 (en) 1996-08-28 2002-03-05 President And Fellows Of Harvard College Detection of multiple nucleic acid sequences in a fluid sample
ATE243761T1 (de) 1996-10-04 2003-07-15 Dako As Sonden zur detektion von mycobakterien
US6027945A (en) 1997-01-21 2000-02-22 Promega Corporation Methods of isolating biological target materials using silica magnetic particles
US5888778A (en) 1997-06-16 1999-03-30 Exact Laboratories, Inc. High-throughput screening method for identification of genetic mutations or disease-causing microorganisms using segmented primers
AU8572898A (en) 1997-07-17 1999-02-10 Tm Technologies, Inc. Methods and compositions for modulating melting temperatures of nucleic acids
US20050037397A1 (en) 2001-03-28 2005-02-17 Nanosphere, Inc. Bio-barcode based detection of target analytes
US6673914B1 (en) 1998-01-22 2004-01-06 John Wayne Cancer Institute Human tumor-associated gene
US20020187470A1 (en) 1998-06-24 2002-12-12 Casey Warren Michael Detection of single nucleotide polymorphisms
US6306643B1 (en) 1998-08-24 2001-10-23 Affymetrix, Inc. Methods of using an array of pooled probes in genetic analysis
US6268147B1 (en) 1998-11-02 2001-07-31 Kenneth Loren Beattie Nucleic acid analysis using sequence-targeted tandem hybridization
US6261779B1 (en) * 1998-11-10 2001-07-17 Bio-Pixels Ltd. Nanocrystals having polynucleotide strands and their use to form dendrimers in a signal amplification system
US6670464B1 (en) 1998-11-17 2003-12-30 Curagen Corporation Nucleic acids containing single nucleotide polymorphisms and methods of use thereof
CA2386239A1 (fr) 1999-10-04 2001-04-12 University Of Medicine And Dentistry Of New Jersey Methodes d'identification de composes liant un arn
US6428957B1 (en) 1999-11-08 2002-08-06 Agilent Technologies, Inc. Systems tools and methods of assaying biological materials using spatially-addressable arrays
US6727356B1 (en) 1999-12-08 2004-04-27 Epoch Pharmaceuticals, Inc. Fluorescent quenching detection reagents and methods
US7955794B2 (en) 2000-09-21 2011-06-07 Illumina, Inc. Multiplex nucleic acid reactions
WO2001083814A2 (fr) 2000-05-04 2001-11-08 Syngenta Participations Ag Nouvelles methodes d'analyse d'acides nucleiques
US7033758B2 (en) 2000-06-02 2006-04-25 Bayer Corporation Highly sensitive gene detection and localization using in situ branched-DNA hybridization
US7439016B1 (en) 2000-06-15 2008-10-21 Digene Corporation Detection of nucleic acids by type-specific hybrid capture method
US6803200B2 (en) 2000-12-12 2004-10-12 Invitrogen Corporation Compositions and methods for the release of nucleic acid molecules from solid matrices
US20020106644A1 (en) 2001-02-05 2002-08-08 Carsten Rosenow Methods for transcription detection and analysis
AU2002252370A1 (en) 2001-03-12 2002-09-24 Irm, Llc. Genomics-driven high speed cellular assays, development thereof, and collections of cellular reporters
US20040161741A1 (en) 2001-06-30 2004-08-19 Elazar Rabani Novel compositions and processes for analyte detection, quantification and amplification
CA2459320C (fr) 2001-09-06 2016-07-05 Genomic Profiling Systems, Inc. Detection rapide de cellules de replication
US20030165935A1 (en) 2001-11-21 2003-09-04 Vann Charles S. Digital assay
US20040023248A1 (en) 2001-12-07 2004-02-05 Whitehead Institiute For Biomedical Research Methods and reagents for improving nucleic acid detection
ES2282599T3 (es) 2002-01-07 2007-10-16 Norchip A/S Metodo para detectar arnm del virus del papiloma humano.
US20040115686A1 (en) 2002-05-17 2004-06-17 Douglas Dolginow Materials and methods to detect alternative splicing of mrna
WO2004020654A2 (fr) 2002-08-30 2004-03-11 Bayer Healthcare Llc Dosages d'acide nucleique en rapport avec une phase solide, combinant une haute affinite avec une haute specificite
US8394944B2 (en) 2002-09-20 2013-03-12 Siemens Healthcare Diagnostics Inc. Dual-purpose primers and probes for providing enhanced hybridization assays by disruption of secondary structure formation
US7122384B2 (en) 2002-11-06 2006-10-17 E. I. Du Pont De Nemours And Company Resonant light scattering microparticle methods
WO2005054516A2 (fr) 2003-11-26 2005-06-16 Advandx, Inc. Sondes d'acides nucleiques peptidiques pour l'analyse de certaines especes staphylococcus
US7462475B2 (en) 2004-05-20 2008-12-09 Dna Poleymerase Technology, Inc. Use of whole blood in PCR reactions
CA2572338A1 (fr) 2004-06-24 2006-01-05 Veridex, Llc. Procedes et reactifs pour la detection de melanome
US8063196B2 (en) 2005-02-01 2011-11-22 Siemens Healthcare Diagnostics Inc. Highly orthogonal universal sequences for use in nucleic acid assays
US7524631B2 (en) 2005-02-02 2009-04-28 Patterson Bruce K HPV E6, E7 mRNA assay and methods of use thereof
US8632970B2 (en) 2005-05-09 2014-01-21 Affymetrix, Inc. Multiplex capture of nucleic acids
CA2606723A1 (fr) 2005-05-09 2006-11-16 Panomics, Inc. Capture multiplex d'acides nucleiques
EP1880025B1 (fr) 2005-05-12 2011-03-16 Affymetrix, Inc. Essais réalisés sur de l'adn à chaîne ramifiée multiplexe
US20090081688A1 (en) 2005-06-20 2009-03-26 Advanced Cell Diagnostics Methods of detecting nucleic acids in individual cells and of identifying rare cells from large heterogeneous cell populations
WO2007044427A2 (fr) 2005-10-05 2007-04-19 Panomics, Inc. Détection d’acides nucléiques provenant de sang total
ATE553211T1 (de) 2005-11-10 2012-04-15 Affymetrix Inc Nachweis von nukleinsäuren über amplifikation von stellvertreternukleinsäuren
JP2008043332A (ja) 2006-08-17 2008-02-28 Panomics Inc 組織スライドからの核酸定量
US7615351B2 (en) 2006-12-01 2009-11-10 Panomics, Inc. Two stage nucleic acid amplification using an amplification oligomer
US20090298709A1 (en) 2008-05-28 2009-12-03 Affymetrix, Inc. Assays for determining telomere length and repeated sequence copy number
US20100081131A1 (en) 2008-10-01 2010-04-01 Ach Robert A Identification of microbes using oligonucleotide based in situ hybridization
US8715926B2 (en) 2008-11-24 2014-05-06 Loma Linda University Biomarkers for the detection of head and neck tumors
WO2010080566A1 (fr) 2008-12-18 2010-07-15 Siemens Healthcare Diagnostics Inc. Procédés et réactifs pour raccourcir les temps d'incubation dans des dosages par hybridation
WO2010129941A1 (fr) 2009-05-08 2010-11-11 Becton, Dickinson And Company Corrélation de l'expression des protéines e6 et e7 de pvh avec la progression de la maladie du col de l'utérus
US20130023433A1 (en) 2009-09-28 2013-01-24 Yuling Luo Methods of detecting nucleic acid sequences with high specificity
ES2719502T3 (es) 2010-01-29 2019-07-10 Advanced Cell Diagnostics Inc Métodos de detección in situ de ácidos nucleicos
CA2787487C (fr) * 2010-02-26 2018-07-24 Ventana Medical Systems, Inc. Sondes polytag
US20120003648A1 (en) 2010-07-01 2012-01-05 Affymetrix, Inc. Signal Multiplexing and Signal Amplification
US20120052498A1 (en) 2010-07-01 2012-03-01 Affymetrix, Inc. Detection of Nucleic Acids
US20120004132A1 (en) 2010-07-02 2012-01-05 Affymetrix, Inc. Detection of Nucleic Acids and Proteins
WO2012040168A2 (fr) 2010-09-20 2012-03-29 Advanced Cell Diagnostics, Inc. Marqueurs biologiques permettant de différencier un mélanome d'un nævus bénin sur la peau
CN104849472A (zh) 2010-10-21 2015-08-19 领先细胞医疗诊断有限公司 用于原位检测核酸的超灵敏方法
US20120172246A1 (en) 2010-12-31 2012-07-05 Affymetrix, Inc. Detection of Nucleic Acids
US20120178081A1 (en) 2010-12-31 2012-07-12 Affymetrix. Inc. Methods of Labeling Cells, Labeled Cells, and uses Thereof
DK2668290T3 (en) 2011-01-28 2017-09-18 Advanced Cell Diagnostics Inc RNASCOPE® HPV ASSAY FOR DETERMINING HPV STATUS OF CANCER AND HEALTH CANCER AND LESSIONS
EP2823066A4 (fr) 2012-03-06 2015-11-11 Advanced Cell Diagnostics Inc Test chromogène double pour la détection in situ d'acides nucléiques
US20140178869A1 (en) 2012-04-05 2014-06-26 Advanced Cell Diagnostics, Inc. Detection of immunoglobulin light chain restriction by rna in situ hybridization
US20130294826A1 (en) 2012-05-04 2013-11-07 Advanced Cell Diagnostics, Inc. Lock-in slide rack
CN105247077A (zh) 2013-03-28 2016-01-13 领先细胞医疗诊断有限公司 通过原位杂交区分瞬时高危型hpv感染和持续高危型hpv感染

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4366241B1 (fr) 1980-08-07 1988-10-18
EP0795610A1 (fr) * 1996-03-13 1997-09-17 Becton, Dickinson and Company Amplification de signal d'hybrisation "in situ" par dépÔt de tyramine biotinilée
WO2007002006A2 (fr) * 2005-06-20 2007-01-04 Panomics, Inc. Detection multiplex d’acides nucleiques
WO2007001986A2 (fr) * 2005-06-20 2007-01-04 Yuling Luo Methodes de detection des acides nucleiques dans des cellules individuelles et d'identification de cellules rares a partir de grandes populations cellulaires heterogenes
US7709198B2 (en) 2005-06-20 2010-05-04 Advanced Cell Diagnostics, Inc. Multiplex detection of nucleic acids

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BOBROW M N ET AL: "UNIT 8.9 Tyramide Signal Amplification (TSA) Systems for the Enhancement of ISH Signals in Cytogenetics", CURRENT PROTOCOLS IN CYTOMETRY, JOHN WILEY & SONS, INC, US, no. supplement 11, 1 January 2000 (2000-01-01), pages 8.9.1 - 8.9.16, XP002477763, ISSN: 1934-9297 *
DEICHMANN M ET AL: "Ultra-sensitive FISH is a useful tool for studying chronic HIV-1 infection", JOURNAL OF VIROLOGICAL METHODS, ELSEVIER BV, NL, vol. 65, no. 1, 1 April 1997 (1997-04-01), pages 19 - 25, XP026831788, ISSN: 0166-0934, [retrieved on 19970401] *
LIZARD G ET AL: "IN SITU HYBRIDIZATION DETECTION OF SINGLE-COPY HUMAN PAPILLOMAVIRUS ON ISOLATED CELLS, USING A CATALYZED SIGNAL AMPLIFICATION SYSTEM: GENPOINT", DIAGNOSTIC CYTOPATHOLOGY, NEW YORK, NY, US, vol. 24, no. 2, 1 February 2001 (2001-02-01), pages 112 - 116, XP009053918, ISSN: 8755-1039, DOI: 10.1002/1097-0339(200102)24:2&LT,112::AID-DC1020&GT,3.0.CO,2-6 *

Also Published As

Publication number Publication date
CN107365847A (zh) 2017-11-21
ES2648564T3 (es) 2018-01-04
US20120100540A1 (en) 2012-04-26
US20140249040A1 (en) 2014-09-04
CN103429755A (zh) 2013-12-04
WO2012054795A1 (fr) 2012-04-26
EP2630260A1 (fr) 2013-08-28
US8658361B2 (en) 2014-02-25
ES2562821T3 (es) 2016-03-08
EP3034625B1 (fr) 2017-10-04
US20160201117A1 (en) 2016-07-14
EP2630260B1 (fr) 2015-11-18
CN104849472A (zh) 2015-08-19
US9315854B2 (en) 2016-04-19

Similar Documents

Publication Publication Date Title
EP3034625B1 (fr) Procédé ultrasensible de détection in situ d'acides nucléiques
US20200399689A1 (en) Methods of detecting nucleic acid sequences with high specificity
US20210032689A1 (en) Methods of detecting nucleic acids in individual cells and of identifying rare cells from large heterogeneous cell populations
US20230128368A1 (en) Set of Oligonucleotide Probes as Well as Methods and Uses Thereto
US20230151413A1 (en) Methods of detecting nucleic acids in individual cells and of identifying rare cells from large heterogeneous cell populations
EP2529030B1 (fr) Methode de detection d'acides nucleiques in situ
EP3561071B1 (fr) Analyses d'expression génique effectuées par analyse élémentaire
Cassidy et al. Developments in in situ hybridisation
JP5272063B2 (ja) イン・サイチュー解析のためのオリゴヌクレオチドプローブおよびタンパク質を標識するためのオリゴヌクレオチド
US20150045251A1 (en) Duplex chromogenic assay for in situ detection of nucleic acids
US20160046984A1 (en) Robust Detection of Nucleic Acids in Situ
JP2005503753A5 (fr)

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 2630260

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ADVANCED CELL DIAGNOSTICS, INC.

17P Request for examination filed

Effective date: 20161209

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20170509

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

AC Divisional application: reference to earlier application

Ref document number: 2630260

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 934092

Country of ref document: AT

Kind code of ref document: T

Effective date: 20171015

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011042230

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2648564

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20180104

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 934092

Country of ref document: AT

Kind code of ref document: T

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180104

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180105

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180204

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180104

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602011042230

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171021

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171031

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171031

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20171031

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171031

26N No opposition filed

Effective date: 20180705

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171021

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20171021

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20111021

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602011042230

Country of ref document: DE

Representative=s name: GLAWE DELFS MOLL PARTNERSCHAFT MBB VON PATENT-, DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20171004

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230516

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20231026

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231027

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20231102

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20231023

Year of fee payment: 13

Ref country code: FR

Payment date: 20231025

Year of fee payment: 13

Ref country code: DE

Payment date: 20231027

Year of fee payment: 13